You are on page 1of 8

SACKLER SPECIAL FEATURE: PERSPECTIVE

Architecture of human telomerase RNA


Qi Zhang, Nak-Kyoon Kim, and Juli Feigon1
Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095-1569
Edited by Neal F. Lue, Weill Cornell Medical College, New York, NY, and accepted by the Editorial Board July 22, 2011 (received for review June 8, 2011)

Telomerase is a unique reverse transcriptase that catalyzes the addition of telomere DNA repeats onto the 3′ ends of linear chromosomes
and plays a critical role in maintaining genome stability. Unlike other reverse transcriptases, telomerase is unique in that it is a ribonu-
cleoprotein complex, where the RNA component [telomerase RNA (TR)] not only provides the template for the synthesis of telomere DNA
repeats but also plays essential roles in catalysis, accumulation, TR 3′-end processing, localization, and holoenzyme assembly. Biochemical
studies have identified TR elements essential for catalysis that share remarkably conserved secondary structures across different species as
well as species-specific domains for other functions, paving the way for high-resolution structure determination of TRs. Over the past
decade, structures of key elements from the core, conserved regions 4 and 5, and small Cajal body specific RNA domains of human TR have
emerged, providing significant insights into the roles of these RNA elements in telomerase function. Structures of all helical elements of
the core domain have been recently reported, providing the basis for a high-resolution model of the complete core domain. We review
this progress to determine the overall architecture of human telomerase RNA.

Box H/ACA RNA | NMR | pseudoknot | telomerase reverse transcriptase

T
elomerase is a large, multisubunit conserved across different species, and it TRs across different species, which in-
ribonucleoprotein (RNP) that usually contains four major functional cludes a large loop containing the tem-
replicates the 3′ end of linear domains: the TERT N-terminal domain plate, a 5′ template boundary element,
chromosomes by processive syn- (TEN), the TERT RNA binding domain a pseudoknot, a loop-closing helix, and
thesis of telomere DNA repeats. Telo- (TRBD), the reverse transcriptase domain a stem terminus element (STE) (9, 15, 46,
meres, the physical ends of linear (RT), and the C-terminal extension (27, 48). The conservation of secondary struc-
chromosomes, generally comprise dsDNA 30–32). The TEN domain interacts with tures rather than sequences suggests a role
with a short repeating species-specific se- the ss telomere DNA repeats, the TRBD for these RNA structural motifs in telo-
quence ending in a 3′ single-stranded domain binds multiple sites of TR, and the merase function, and indeed, these regions
overhang of variable length plus associated RT and C-terminal extension domains of TR are essential for synthesis of telo-
telomere binding proteins, called shelterin bind the RNA/DNA hybrid and catalyze mere repeats (49–55). Other regions of
in humans (1, 2). Telomeres protect the the addition of DNA repeats onto the 3′ TR are involved in species-specific roles in
integrity of linear chromosomes by allow- end (27, 31–34). Although no structures of telomerase biogenesis, RNA processing,
ing the cellular DNA repair machinery TERT with all four domains have yet been localization, and accumulation (56–61).
to distinguish them from double-strand reported for any species, structures of the A conserved secondary structure for
breaks, thus playing critical roles in main- TEN and TRBD domains from Tetrahy- vertebrate TRs was first determined on the
taining genome stability in eukaryotes (1, mena thermophila telomerase (33, 35) and basis of 35 sequences (46), with four pro-
2). Shortening of telomeres below a criti- the full-length Tribolium castaneum TERT posed conserved structural domains: (i)
cal length because of inherent incomplete that lacks the TEN domain have been re- the pseudoknot, which includes the tem-
replication of DNA ends ultimately leads ported (36, 37). These high-resolution plate, and (ii) the conserved regions 4 and
to telomere fusions and cell senescence structures have significantly advanced our 5 (CR4/CR5), which together comprise
(3–6). The 3′ ends of telomeres are repli- understanding of how TERT catalyzes the the catalytic core of the TR; (iii) the box
cated by telomerase, a unique reverse reverse transcription of telomere DNA
H/ACA; and (iv) the CR7. An updated
transcriptase discovered almost three and how TERT could potentially interact
secondary structure of hTR (47, 62) is
decades ago (7), which catalyzes the ad- with the ss telomere DNA and the tem-
shown in Fig. 1 with known associated
dition of telomere DNA repeats onto the plate RNA/DNA hybrids. Excellent re-
holoenzyme proteins. All of the secondary
ends of linear chromosomes using an views can be found elsewhere that describe
structure elements found in common
embedded RNA as the template (8, 9). the current state of knowledge about the
Although telomerase has a low or structure and function of TERT (31, 32). among TRs across different species are in
undetectable level of activity in most so- Although the essential templating the 5′ region of hTR, where the STE is the
matic cells, it is active in some germline, function of TR was discovered more than
epithelial, and hematopoietic cells, and it 20 y ago (38), the TR contains more than
is highly active in the majority (∼90%) of a template. To date, the TR sequences of This paper results from the Arthur M. Sackler Colloquium
of the National Academy of Sciences, “Telomerase and
cancer cell lines (10–12). Telomerase de- 28 ciliates, 43 vertebrates, and 25 yeasts Retrotransposons: Reverse Transcriptases That Shaped Ge-
ficiency because of mutations in human have been determined (39–41). In contrast nomes” held September 29–30, 2010, at the Arnold and
telomerase RNA (hTR) has also been to the relatively conserved TERT, TRs Mabel Beckman Center of the National Academies of Sci-
linked to several inherited human dis- differ greatly not only in sequence but also ences and Engineering in Irvine, CA. The complete pro-
gram and audio files of most presentations are available
eases, such as dyskeratosis congenita, in length, ranging from 147 to 205 nt in on the NAS Web site at www.nasonline.org/telomerase_
aplastic anemia, myelodysplasia, and idio- ciliates (38, 42–45), from 312 to 559 nt and_retrotransposons.
pathic pulmonary fibrosis (13–26). in vertebrates (46, 47), and from 779 to Author contributions: Q.Z., N.-K.K., and J.F. wrote the
The telomerase holoenzyme includes >2,030 nt in yeasts (40, 41). Despite sig- paper.
a unique reverse transcriptase [telomerase nificant challenges in identifying common The authors declare no conflict of interest.
reverse transcriptase (TERT)], an essential functional TR elements because of such This article is a PNAS Direct Submission. N.F.L. is a guest
RNA (TR), and several species-specific divergence, phylogenetic and mutational editor invited by the Editorial Board.
proteins required for proper function in studies have revealed a conserved sec- 1
To whom correspondence should be addressed. E-mail:
vivo (27–29). The protein TERT is highly ondary structure found in common among feigon@mbi.ucla.edu.

www.pnas.org/cgi/doi/10.1073/pnas.1100279108 PNAS | December 20, 2011 | vol. 108 | no. 51 | 20325–20332


CR4/CR5 domain (nucleotides ∼243 to bp P2/P3 pseudoknot are constrained

Core Domain
∼326), and the remaining motifs comprise by the intervening 24-nt template-
the core domain, also known as the tem- TERT containing loop.
plate/pseudoknot domain or the pseu-

CR4/CR5
doknot/core domain (nucleotide 33–191) Structure and Dynamics of the P2b-P3 Pseu-
(46, 47, 63). These two highly conserved doknot. The P2b-P3 pseudoknot contains
structural domains independently bind almost all of the highly conserved nucleo-
TERT (50, 64) and are the only required tides within the hTR core domain (46).
hTR elements for in vitro reconstitution Early studies of minimal pseudoknots
of catalytically active telomerase with containing these conserved nucleotides
showed that the pseudoknot unfolds to

CAAUCCCAAUC
hTERT (49, 50). The 3′ end of hTR has
been identified as an H/ACA small Cajal TCAB1 a P2b hairpin containing an unusual run of
body (CB) -specific RNA (scaRNA; nu- NHP2 U-U and U-C base pair (77) and that
cleotides ∼211 to ∼237 and ∼334 to 451, NOP10 a two-base mutation in the P3 found in
the upper boundary of the 5′ hairpin has GAR1 some patients with dyskeratosis congenita
not been determined) (59, 62, 65) (Fig. 1), Dyskerin
n (13) destabilizes the pseudoknot. Al-
and it plays essential roles in biogenesis though a conformational switch between
and regulation of telomerase holoenzyme CR7 the completely folded pseudoknot and this
in vivo, including accumulation, 3′-end partially folded hairpin was proposed as
processing, and localization of hTR (58– a molecular switch for template trans-
60, 66). The hTR scaRNA binds two sets location (77, 78), both the solution struc-
of the four evolutionary conserved H/ACA ture of the minimal pseudoknot (discussed
RNP proteins (dyskerin, Gar1, Nop10, and below) and mutational analysis suggest
Nhp2) to form an H/ACA RNP (Fig. 1) that a completely folded pseudoknot is
(67). Structures of single hairpin H/ACA 5’
box H ACA
3’ required for optimal activity of telomerase
RNPs from archaea have provided insight (52, 79). The solution structure of the
H/ACA scaRNA minimal hTR pseudoknot is a compact H-
into the likely placement of proteins on
each hairpin of the hTR scaRNA domain Fig. 1. Secondary structure and known protein type pseudoknot with extensive tertiary
(66, 68–71). The hTR scaRNA also con- components of the human telomerase holoen- interactions between the loop and stem
tains a conserved Cajal body localization zyme. The human telomerase RNA (hTR) contains nucleotides, and it revealed an essential
element (CAB box) (59) in its 3′ terminal three major structural and functional domains, triple helix in the pseudoknot (52). The
hairpin loop in the region identified as the the core domain, the CR4/CR5 domain, and the H/ functional importance of the tertiary in-
ACA scaRNA domain (46, 59, 62). The hTR core and teractions, described below, was shown by
CR7 domain, identifying it as a scaRNP.
CR4/CR5 domains independently bind the hTERT
The protein telomerase Cajal body protein comparison of thermodynamic stabilities
(blue ellipse) (50, 64). The hTR scaRNA domain
1 (TCAB1)/WD-repeat domain 79 binds two sets of the four H/ACA RNP proteins:
of pseudoknots with mutations and com-
(WDR79) that binds the CAB box and dyskerin (green), Gar1 (cyan), Nop10 (magenta), pensatory mutations and the effects of
drives localization of hTR into Cajal body and Nhp2 (orange) (67). The protein TCAB1/ the same nucleotide substitutions in-
was recently identified (72, 73). WDR79 (purple) binds both the dyskerin and the corporated into the full-length TR on te-
To date, there are no crystal structures of CAB box located at the CR7 region within the H/ lomerase activity in vitro (52). The tertiary
telomerase RNA or telomerase protein– ACA scaRNA domain (72, 73). structure of the minimal hTR pseudoknot
RNA complexes; however, over the last also provided a structural explanation for
several years, structures of several key the effect of disease mutations in this re-
hTR elements have been determined by pseudoknot, all four helices (P2a.1, P2a, gion of hTR (52, 54). Mutational studies
NMR spectroscopy (52, 54, 55, 60, 74–76). P2b, and P3) have been shown to be re- and modeling of yeast telomerase pseu-
This review summarizes what NMR quired for telomerase activity, and disease- doknot domains (53, 80) have provided
structural and dynamics studies, combined linked mutations disrupting these helical additional evidence for an essential role
with biochemical and mutational analysis, structures severely impair telomerase ca- for a conserved triple helix in the pseu-
have revealed about the functional roles of talysis (63). Between these helical regions doknot domain. The putative pseudoknots
hTR domains and the overall architecture are internal loop regions. Except for loop in ciliates are much smaller than those
of hTR. J2b/3 and the 3′ portion of loop J2a/3, pseudoknots in yeasts and vertebrates.
which are critical for the formation of P2b- Although no structures of ciliate pseu-
Core Domain of hTR P3 pseudoknot, most of these loop regions doknots have yet been reported, sequence
The core domain has been the main focus are not conserved, and their nucleotide analysis and modeling of 28 ciliate TRs
of biochemical and biophysical studies of identities are not essential for telomerase have predicted one (and in one case, two)
hTR, because it contains most of the activity. The P2b-P3 pseudoknot is linked potential base triples in the ciliate pseu-
conserved nucleotides and most of the to the P1 helix through three nucleotides doknots (81).
disease-linked mutations (21–24, 26, 48, at its 3′ end and to the P2a helix through In the minimal hTR pseudoknot
62). It is the largest functional RNA do- an asymmetric bulge J2a/b at its 5′ end (PKDU), the bulge U177 was removed to
main at the 5′ end of hTR. Biochemical (Fig. 2A). On the other side of the P2a stabilize the pseudoknot formation. How-
characterizations have identified nucleo- helix are P2a.1 and the adjacent internal ever, deletion of this residue in hTR
tides between residues 33 and 191 as the loop J2a.1, which are a mammalian-spe- resulted in a 2- to 10-fold decrease in
minimal hTR core domain required for cific extension to P2a helix (Fig. 2A). The telomerase activity (52, 78), and compu-
telomerase catalytic activity in vitro (63). 5′ end of the P2a.1 helix and 3′ end of the tational modeling of the P2b-P3 pseu-
This region can be further divided into P1 helix are linked by the large template- doknot suggested that deletion of U177
three major segments, a large loop con- containing loop. These secondary struc- alters tertiary interactions within the
taining the template, the P1 helix as the ture features suggest a unique and overall pseudoknot (82). In addition, substitution
loop-closing helix, and a full-length P2/P3 compact architecture adopted by the hTR of the 2′-OH of residue A176, which is
pseudoknot (Fig. 2A). Within the P2/P3 core domain, because the ends of the ∼43- adjacent to residue U177, with 2′-OMe or

20326 | www.pnas.org/cgi/doi/10.1073/pnas.1100279108 Zhang et al.


A 60 G
A A
UCC
U
C
C
G
G
G
A
C
B C D
A G C

P2a.1
CC G C
G G U G
U A U 5’
U C G U
C
G
G
C C 5’
C G

J2a.1
G A
A C C
C U
U G U
A C

J2a/3
U160 C
U C G
G C
P2a U
Template

A U A
C A U
A U G
C
J2a/b

A U A
CG G
C U
CCC
G C
A G C A
C G A U177
A G C
P2b

G G A
U G120C C
C G
G C A
C A U A
C GA
U U A
U
U A
100 U 5’
G U AU177
U
C G
U U
A U
J2b/3

G C P3
U
U U A
C
C G U
U CG C 5’
U
40U U G
U A CU
3’ 3’ 3’
C G 3’
C G
G C P1
G C
5’ 3’
P2b-P3 PKDU P2b-P3 PKWT P2a-J2a/b-P2b P2a-J2a.1-P2a.1

Fig. 2. Structures of subdomains of the hTR core domain. (A) Sequence and secondary structure of the hTR core domain, which can be divided into four
subdomains: P2a-J2a.1-P2a.1 (blue-gray-gold), P2a-J2a/b-P2b (gold-green-red), P2b-P3 pseudoknot (red-pink), and P1 (dark green) (46). (B) NMR solution
structures of the P2b-P3 pseudoknot, where PKDU (PDB ID 2K96) and PKWT (PDB ID 2K96) are structures for ΔU177 mutant and WT constructs, respectively
(54). (C) NMR solution structure of P2a-J2a/b-P2b (PDB ID 2L3E) (55). (D) Structural model of the P2a-J2a.1-P2a.1 determined by the RDC-MC-Sym approach (55).
All 3D structures are color-coded like the secondary structure in A, and nonnative residues are colored light gray.

2′-H results in about a twofold decrease in a junction that is a Hoogsteen base pair helical interactions. Based on the struc-
activity, leading to the proposal that the formed between the first nucleotide (U99) ture, the base of U177 sits right over the
A176 2′-OH may contribute directly to in the J2b/3 loop and the last nucleotide 2′-OH of residue A176 and would steri-
telomerase catalysis (53). To provide (A173) in the J2a/3 loop, and both pseu- cally block its accessibility during telo-
structural insight into the WT P2b-P3 doknots are stabilized by the same base merase catalysis (Fig. 3D). Dynamic
pseudoknot (PKWT) and the functional triples flanking this junction base pair characterization by NMR spin relaxation
role of U177, the solution structure of (Figs. 2B and 3B). The A-rich J2a/3 loop measurements revealed that U177 is in-
PKWT was determined, the original forms two minor groove base triples with trinsically highly flexible, which suggests
structure of PKDU was further refined the P2b helix, and the U-rich J2b/3 loop that, rather than blocking A176, U177 may
with an extensive set of NMR residual forms three major groove U-A·U Hoogs- serve as a hinge to provide additional
dipolar couplings (RDCs) (Fig. 2B), and teen base triples with P3 helix (Fig. 3C). backbone flexibility for residue A176 to
systematic structural and dynamical com- However, PKDU is also stabilized by an facilitate the catalysis (54).
parisons were made between PKWT and additional C112·G178-U103 triple that is The structure determination of PKWT
PKDU (54). These structural character- disrupted in PKWT because of the pres- also revealed why it is thermodynamically
izations revealed that PKWT folds into the ence of U177, which creates a large roll less stable than PKDU (52). Comparison
same H-type pseudoknot conformation as and tilt between the flanking base pairs of NMR imino proton spectra of PKWT
PKDU with almost identical tertiary in- (Fig. 3 A and D). In PKWT, U177 is flip- and PKDU as a function of temperature
teractions (Figs. 2B and 3 A and B). In ped out of the P3 helix into the minor revealed an unanticipated difference in
both PKWT and PKDU, the P2a and P2b groove and is located on the opposite side the late folding/early unfolding pathway.
helices are stacked on either side of of the helix from the major groove triple In PKDU, as expected, when temperature
increases, the Hoogsteen base pairs be-
tween the J2b/3 loop and P3 helix begin to
A A171
B C melt before the Watson–Crick base pairs
3’ 5’ 5’ in P3 helix. However, in PKWT, the
G118 C96 C170
U
U97 A172 A117 U97
presence of bulge U177 results in partial
A171
A117 U unstacking of the three Watson–Crick U-
C116 G98 A172
G98 A173 A base pairs from the rest of the helix
C116 U100 A173 U99 A below the bulge U177 (Fig. 3A). As a
U115
U115 A174 U100 consequence, the three U-A base pairs
A174
U114 U114 A175 U101 D above and the two base pairs below the
A175
U113A176 U101 U113 A176 U102 A176 bulge U are thermodynamically less stable
U113
U102 U17
7 U1 than the tertiary (Hoogsteen base pair)
C112 G178 03 C112
C112 interactions, and they begin melting before
A111 U179
G178 C104 the Hoogsteen U-A base pairs. Thus, the
U103 G110 C180 G178
U177 5’ presence of U177 in PKWT results in an
3’ 3’
altered late folding/early unfolding path-
way in the pseudoknot, which may be rel-
Fig. 3. Tertiary interactions in the pseudoknot. (A) The structure of the triple helical region of PKWT
(54). (B) Schematic representations of the tertiary interactions in PKWT. In PKDU, U103 forms a Hoogs-
evant to RNA assembly. In addition, for
teen base pair with G178. (C) U·A-U Hoogsteen base triple. (D) Detailed view of the base pairs sur- both pseudoknots, the melting studies re-
rounding U177. The base of U177 stacks over the 2′OH of A176. The nucleotides are in CPK colors, except vealed that the junction J2b/3 loop–J2a/3
that U177 is colored magenta. loop Hoogsteen U-A base pair is

Zhang et al. PNAS | December 20, 2011 | vol. 108 | no. 51 | 20327
remarkable stable. Thus, this base pair relative to each other with a motional motion between P2a.1 and P2a is re-
plays a critical role in overall folding of the amplitude of ∼39°. In contrast, HIV-1 markably restricted (∼26° cone motions)
pseudoknot (54). TAR RNA has a cone-model motional based on dynamic characterization by
amplitude of ∼55° (91). NMR RDCs. The stability between
Structure, Dynamics, and Function of the P2a- The rare structure and unexpected dy- helices is consistent with the observations
J2a/b-P2b. Adjacent to the highly conserved namics of J2a/b suggested that it might that disruption of base pairs flanking
P2b-P3 pseudoknot is a 5-nt bulge loop, have an important functional role in telo- J2a.1 decreases telomerase activity and
J2a/b, which serves as a bridge between the merase RNA topology and function. To that nonmammalian vertebrate TRs
pseudoknot and the P2a helix. This py- test this hypothesis, systematic mutations have a single long P2a helix without a
rimidine-rich J2a/b bulge loop is not highly were carried out to investigate the effects J2a.1 (46, 47).
conserved in sequence, except for a rela- of the length, strand location, and sequence
tively conserved G at the 5′ end (61% in of J2a/b on telomerase function (55). The High-Resolution Model of the hTR Core Do-
vertebrate and 83% in mammalian TRs) results showed not only that the di- main. The structures and dynamic anal-
(39). Initial studies showed that swapping rectional bending defined by J2a/b is re- ysis of subdomains of the hTR core do-
the bulge sequence 5′→3′ or replacing the quired for overall telomerase activity but main, in particular, the conserved
hTR sequence with the mouse TR se- also that the intrinsic flexibility across J2a/ pseudoknot, have provided great insights
quence had little effect on human telo- b is important for processive catalysis by into their functional roles in telomerase
merase catalytic activity (63, 83). These telomerase. The results also suggest the catalysis. However, they do not provide an
results, along with the lack of sequence consensus sequence G83Y78Y87Y96Y87 for overview of how the P2/P3 pseudoknot
conservation, argued against an important the 5-nt bulge in mammalian TRs may folds and how its architecture imposes
role of the J2a/b bulge loop in telomerase have evolved for both nucleotide addition conformational constraints that position
activity. However, the location of the and template translocation during telo- the template into the active site of telo-
J2a/b bulge loop is conserved in all verte- merase catalysis. Finally, the large bend at merase. A low-resolution (6.5–8.0 Å)
brate TRs, and its length is usually 5 nt in the J2a/b bulge would be expected to play FRET model of the hTR core domain has
mammalian TRs (46, 47). The solution a significant role in determining the over- been reported (PDB ID 2INA), where
structure of the J2a/b bulge loop together all topology of the core domain, which distances derived from FRET between
with flanking P2a and P2b helices showed discussed more below. fluorescently labeled peptide nucleic acids
that J2a/b introduces a large bend (89 ± were used in structural modeling (93). In
3°) between P2a and P2b across the major Structure and Dynamics of the Mammalian- this approach, although these distances
groove (Fig. 2C) (55). The 5′-end residue Specific P2a.1-J2a.1-P2a. Located on the were obtained in the full-length hTR, they
of the bulge loop, G84, stacks above P2a, other side of hTR P2/P3 pseudoknot is the provide only indirect constraints on the
and the 3′-end residue of the bulge loop, P2a.1-J2a.1-P2a domain, where P2a.1 is positions of the helical regions of the hTR
C88, stacks below P2b. A change in the a mammalian-specific extension to the P2a core domain, because the peptide nucleic
backbone direction occurs at the center helix through an asymmetric internal loop acids were hybridized onto three single-
bulge residue, U86, which leads to an J2a.1. Biochemical studies have shown that stranded regions, the 5′ end of hTR (nu-
overall S-shape conformation of J2a/b. some nucleotide substitutions in this region cleotides 1–13), the template region (nu-
Remarkably, the S-shape structure of the and the C72G mutation associated with cleotides 44–56), and the J2a/3 region
J2a/b results in almost no twist between aplastic anemia result in a decrease in (nucleotides 146–158). One caveat is that
P2a and P2b (−10 ± 10°) (Fig. 2C). A telomerase activity (62, 63, 83). In NMR the inherent flexibility of the single-strand
search for other 5-nt bulge structures, us- spectra of a P2a.1-J2a.1-P2a construct, regions, in particular, at the 5′ end of the
ing the FRABASE program (84), un- most of the nonexchangeable protons hTR, was not considered in the modeling.
covered only one other 5-nt bulge among from the J2a.1 loop and surrounding nu- Recently, by applying a computational
all RNA structures solved to date from the cleotides were exchange-broadened and in modeling approach that incorporates the
hepatitis C virus (HCV) internal ribosome many cases, not visible. This finding not Assisted Model Building with Energy Re-
entry site (IRES) domain II (85). Even only indicated the presence of significant finement (AMBER) force field and short-
more surprisingly, this HCV IRES domain conformational exchange in J2a.1, appar- range NOE restraints from individual
II bulge adopts the same S-shaped struc- ently because of the bases in the internal structures, a high-resolution structure
ture, despite the fact that it has a different loop adopting more than one conforma- model of the P2/P3 pseudoknot was de-
sequence from the hTR J2a/b. A some- tion involving alternative base pairs, but termined that contains all nucleotides ex-
what broader search that also allowed for also hindered high-resolution structure cept for the nonconserved single-strand
noncanonical closing base pairs and determination of this region of hTR by region of J2a/3 (Fig. 4A) (55). There are
swapping the strand with the 5-nt bulge solution NMR (55). To overcome these significant differences between the FRET-
uncovered two additional sequences, and difficulties, the RDC-MC-Sym approach and NMR-based structure models in both
these sequences have similar S-shaped in- was developed for structure determination the local structures and relative helical
ternal loops (86, 87). Thus, the J2a/b bulge of nucleic acids (55). This approach uses orientations, but both models do exhibit
represents a rare structural motif. a combination of computational modeling an open architecture of the hTR core do-
Systematic dynamic characterizations by by program MC-Sym (92) and experi- main and a major interhelical bend oc-
NMR 15N spin relaxation (88) and NMR mentally derived restraints by NMR curring across the J2a/b bulge (55, 93).
RDCs (89, 90) also revealed another un- RDCs. The structural analysis revealed The NMR-based structure model
usual feature of the J2a/b bulge loop. It that P2a.1 is essentially a linear extension revealed that the full-length P2/P3 pseu-
was initially expected that the J2a/b bulge of the P2a helix (Fig. 2D). The interhelical doknot of the hTR core domain folds into
loop would be highly flexible, but although bend between P2a.1 and P2a is only 6 ± 3°, an overall V-shape conformation that is
J2a/b exhibits some flexibility, its motion is and the twist between the two helices is defined by the ∼90° bend across the J2a/
remarkably limited on the nanosecond to 135 ± 14°, which agrees well with the b bulge. The P2b-P3 pseudoknot and
millisecond time scale compared with the amount of twist for an ∼4-bp irregular P2a.1-J2a.1-P2a domain are located on
3-nt bulge loop in HIV-1 transactivation helix formed by the asymmetric loop J2a.1. either side of the J2a/b bulge, and they are
response element (TAR) RNA. Based on Although J2a.1 undergoes significant each ∼50Å long. Thus, the bend across
a cone motional model, P2a and P2b move conformational exchange, the interhelical J2a/b creates an ∼70 Å end to end

20328 | www.pnas.org/cgi/doi/10.1073/pnas.1100279108 Zhang et al.


A B
Palm Fingers

A176
U177
DNA

Thumb TRBD
A62
Template TEN CP T 1 2 A B’ C D E CTE
Human TERT
Tribolium castaneum TERT
3’ 5’

Fig. 4. Models of the hTR core domain and interaction with TERT. (A) NMR-based model of the hTR core domain including a DNA primer bound to the
template (55). (B) The hTR P2/P3 pseudoknot positioned onto the T. castaneum TERT in two possible orientations, where the hTR P2/P3 pseudoknot lies either
parallel (Left) or perpendicular (Right) to the T. castaneum TERT-telomeric RNA/DNA complex (PDB ID 3KYL) (37). The color scheme for the hTR P2/P3 pseu-
doknot domain is the same as in Fig. 2. Domains of the T. castaneum TERT are colored and labeled as shown, and the RNA template and telomeric DNA are
colored in purple and cyan, respectively. A comparison of the domain structures of the hTERT and T. castaneum TERT is also shown.

distance between P2b-P3 and P2a.1, which hTR, the CR4/CR5 domain is the STE, has been shown to be essential for TERT
agrees well with the length of the in- and it interacts directly with hTERT in- binding and telomerase activity (50, 98).
tervening 24-nt single strand containing dependently from the core domain. The Both the length of the P6.1 helix and the
the template without significant stretching. hTR CR4/CR5 domain contains a three- two conserved loop residues are critical for
Because of the negligible twist between way junction (Fig. 5A), which has also telomerase activity but not for binding of
the P2a and P2b induced by the J2a/ been proposed to form in yeast telomerase TR to TERT (64, 98). The structure of
b bulge loop, the structural model also RNAs (97). Like the core domain, al- P6.1 showed that the first (U306) and the
revealed that the conserved bulge residue though the CR4/CR5 secondary structure last (G310) residues of the 5-nt loop form
U177 (52, 78) and the catalytically im- is conserved, most conserved residues are a U-G wobble pair on top of the A-form
portant 2′OH of residue A176 (53) are localized in one particular region, the P6.1 helix (Fig. 5B) (75). The remaining three
both positioned on the inner surface of the hairpin. Additional highly conserved resi- residues from the loop, U307, G309, and
core domain and face to the 5′ end of the dues are located at the large internal loop G310, are exposed to solvent, with U307
template, where the active site should be that forms a three-way junction between and G309 located on the minor groove
located. The structural model is also con- P6, P6.1, and P5. Although various Wat- side of the loop and G308 located on the
sistent with the close proximity of the son–Crick base pairs can be drawn be- major groove with partial stacking on
end of the pseudoknot to the template tween nucleotides in the three-way U306. The small loop formed by these
needed to obtain maximal activity in an junction, substitutions and compensatory three residues has a well-defined confor-
engineered cis-telomerase (94). In addi- mutations indicate that it is the sequence mation, and this architecture of the loop
tion, the dynamic characterization showed rather than the base pairs that are impor- has been proposed to allow the essential
that interdomain motion between P2a and tant for telomerase activity (98). In addi- loop nucleotides to interact with TERT or
P2b has an amplitude of ∼39° assuming tion, a minimal CR4/CR5 domain TR (75). Intriguingly, gel shift and cross-
a cone motional model, which can be construct, which includes P6.1, all of P6a- linking experiments have shown that iso-
translated into a displacement of ∼28 Å P6b except the terminal hairpin, and lated hTR fragments of P6.1 and the
between the two ends in the full-length P2/ flanking nucleotides but does not include template can directly interact with con-
P3 pseudoknot. This distance agrees well the P5 helix, is sufficient to reconstitute tacts between the P6.1 loop residues and
with the ∼17 Å that the template must telomerase activity in trans with the core the two ends of the template region (99),
translocate through the active site during domain and TERT (Fig. 5A) (76). Thus, it although the biological relevance of this
the synthesis of each TTAGGG telomere is not clear whether this region forms interaction in the context of hTERT re-
repeat. Although the binding sites of a three-way junction in the context of its mains to be established.
TERT on the core domain have not been association with TERT. To date, no Most noncoding RNAs contain modified
identified, the overall shape of the P2/P3 structural information on the CR4/CR5 nucleotides, and hTR seems to be no ex-
pseudoknot fits nicely into the crystal three-way junction has been reported, but ception. Six potential pseudouridine (Ψ)
structure of the TERT from T. castaneum the structures of two helical regions of the modification sites have been identified
(36, 37). The pseudoknot can be modeled hTR STE have been solved (74–76). These within the hTR, two of which are located
onto the TERT either in parallel or per- structures are the essential P6.1 hairpin in the P6.1 loop (76). Pseudouridine is the
pendicular to where the template/primer and the central portion of P6 surrounding most abundant posttranscriptional modi-
fits in the donut hole of the TERT, such the J6 internal loop. fied RNA nucleotide, and it is found in all
that flexing of J2a/b would allow the tem- species (100). The locations of pseudour-
plate to be pulled through the active site Structures of P6.1 and Pseudouridylated P6.1 idines are usually highly conserved, and
during telomere synthesis (Fig. 4B). This The P6.1 is a short hairpin off of the three- they play important roles in biological
finding provides a testable model for how way junction that has a helix of four functions. To investigate the potential
the core domain interacts with TERT. Watson–Crick base pairs capped by a 5-nt functional roles of these Ψ-modifications
loop. The 13-nt sequence of P6.1 is highly in the essential P6.1 loop, the solution
STE of hTR conserved; in addition to U307 and G309 structure of a pseudouridine-modified
The STE is the other catalytically essential in the loop, all 8 nt forming Watson–Crick P6.1 (Ψ-P6.1) was determined (Fig. 5B)
TR element conserved across ciliates, base pairs are 100% conserved in verte- (76). Furthermore, both the thermody-
vertebrates, and yeasts (9, 50, 95, 96). In brates. The formation of base pairs in P6.1 namic stability of the Ψ-P6.1 vs.

Zhang et al. PNAS | December 20, 2011 | vol. 108 | no. 51 | 20329
A U
C C
G B P6.1 Ψ4P6.1 pseudouridylation may have a subtle but ugAG sequence and serves as a CB local-
U307 Ψ307
C G
U A significant effect on telomerase activity. ization signal (105). Remarkably, the CR7
U G
C G U306 Ψ306 domain of vertebrate TRs, as discussed
P6b G C
A Structure of the P6 Hairpin. The P6 hairpin is below, contains not only a CB localization
270 G C Minimal
G C
G C CR4/CR5
located adjacent to the 5′ end of P6.1. It signal but also another signal for the ac-
J6 C
C
A
C 290
5’
5’
has two helical regions, P6a and P6b, that cumulation and processing of TRs (60).
flank a small asymmetric internal loop J6,
U
C G
P6a G C Although the H/ACA and CR7 domains
G C
P6 and it is capped by a UCCG hairpin (Fig. are not required for telomerase activity
U
C A
260 G C
C U
U C
G 5A). Deletion of the hairpin loop has no
G C
U
U G C G in vitro, they form a scaRNA domain
C G
G C A
G G
G 310
A U P6b
G C
effect on telomerase activity, but addi- that is essential for in vivo accumulation,
C GA G C A
G
C A
C
U
P6.1
C
C U
C 290 tional deletions that include the J6 in- 3′-end processing, and localization of
U C G
A
G
G G C ternal loop have been shown to inhibit
G U G C vertebrate TRs.
G 250 C C interactions between CR4/CR5 and TERT
U G
A U A
P6a Although no high-resolution structures
C
C GC
C 260 G C
and abolish telomerase activity (50).
G C
G C
A U of the H/ACA domain from any verte-
P5 C G G C The solution structure of an RNA de-
C G 5’ 3’ brate TRs with or without H/ACA RNP
C G 5’ rived from P6, including the J6 internal
5’ 3’ proteins have been determined to date,
loop, has been determined (Fig. 5C) (74).
Fig. 5. Structures of subdomains of the hTR CR4/ P6a was predicted on the basis of phylo- a combined structural and biochemical
CR5 domain. (A) Sequence and secondary struc- genetic analysis to have a C262-A295 base study, including localization studies using
ture of the hTR CR4/CR5 domain (46). The minimal pair next to bulge U261, but in the struc- FISH, on the hTR has delineated the
CR4/CR5 required for reconstitution in vitro of ture, a U261-A295 base pair forms and
active telomerase is highlighted by a dashed box
C262 is bulged out. The J6 internal loop
(76). (B) NMR solution structures of P6.1 (PDB ID
1OQ0) (75) and Ψ4P6.1 (PDB ID 2KYE) (76). The
is well-defined, with a potential triple A CR7 B g414
g C C415
formed between C267 through water-

CAB Box
two U and three G letters in the P6.1 loop are A U A413
colored in green and yellow, respectively. The two mediated hydrogen bonds to the G268- g412
U416
C288 base pair. Interestingly, similar to the g g g417
pseudouridines (Ψs) in the P6.1 loop are colored in
red. (C) NMR solution structure and secondary P2a.1-J2a.1-P2a, the interhelical motion u U u411
U418
structure of a P6 hairpin construct (PDB ID 1Z31) 410 C G
between P6a and P6b across this asym- C410
G419
(74). The internal loop residues (C266-C267 and metric bulge is very limited with amplitude C G 420 C409 G420
A289-U291) are colored in green, and the bulge G C
<10°, which was revealed by RDC analysis.
residue C262 is colored in purple. U A 3’ 5’
These structural and dynamical properties 5’ 3’

of J6 result in a unique conformation of


C D

G414
unmodified P6.1 and the effect of pseu- the P6 region. In addition to a 20° inter- C415
helical angle and ∼3 Å deflection between
douridylation on telomerase activity in vi- A413 U416
P6a and P6b, the J6 internal loop forms an
tro were characterized. The loop structure G412
G41
7
unusual solvent-accessible opening, lead-
of the Ψ-P6.1 is significantly different from ing Leeper et al. (74) to speculate that this
U41
1
U418
C410 G419
the loop structure of P6.1. The stem clos- internal loop may be important for TERT C409 G420
ing Ψ306·G310 base pair has a single hy- or TR interactions. However, a variety of C408 G421

drogen bond between the imino proton of base substitutions in this internal loop do 5’
U407 A422
3’
G310 and the O4 of Ψ306, which is dif- not abrogate activity (50), and therefore, it
ferent from the canonical U·G wobble pair E F
G414

is not clear at the current stage that this


found in the unmodified P6.1. The bases structure is unique. C415

of Ψ306 and Ψ307 are located in the major A413 U416

groove of the loop and are stacked on H/ACA scaRNA Domain of hTR G412
G41
7
1
U41
G305 and Ψ306, respectively. These two In vivo biogenesis and regulation of telo- U418
C410 G419
pseudouridines are spatially positioned to merase holoenzyme require additional C409 G420
form water-mediated hydrogen bonds be- telomerase RNA motifs. In vertebrates, the C408 G421

tween the imino protons and their 5′ 3′-half of TR comprises an H/ACA scaR- 5’
U407 A422
3’
phosphate oxygens. Only one base, the NA domain, which includes a telomerase-
nonconserved G308, is flipped out of the specific CR7 domain that forms a 3′-ter- Fig. 6. Structure and function of the hTR CR7
minal hairpin. All H/ACA small nucleolar domain (60). (A) Sequence and secondary struc-
loop, and it is on the minor groove side. ture of the hTR CR7 hairpin and flanking base
The phosphate backbone turns between RNAs (snoRNAs) and scaRNAs form
pairs. Boxed residues are the CAB box sequence.
G308 and G309, and G309 in the syn RNPs with evolutionary conserved H/
Residues with >95% and >85–95% conservation
conformation stacks on the sugar of G310. ACA RNP proteins and generally function among all vertebrate species are shown in capital
to direct site-specific pseudouridylation of letters and bold fonts, respectively. Nonnative
These structural features revealed that the
ribosomal RNAs and small nuclear RNAs,
Ψ-modifications at the loop increase both residues used in structure determination are the
respectively (101–104), but in the case of base pairs below the dashed line. (B) NMR solution
base-stacking and hydrogen-bonding in- telomerase, the domain seems to have structure of the CR7 hairpin (PDB ID 2QH2). Resi-
teractions in the Ψ-P6.1 relative to the been co-opted to help localize telomerase dues are colored by type: orange, A; green, U;
unmodified P6.1, resulting in higher ther- to Cajal bodies (56, 58, 59). The H/ACA blue, G; red, C. (C and D) Residues comprising the
modynamic stability that is characterized motif has a hairpin-hinge-hairpin-tail sec- hTR-specific processing signal are highlighted in
by UV melting studies. Interestingly, the magenta in the secondary structure (C) and col-
ondary structure, where the conserved H
pseudouridine modification at 306 and 307 ored as in B in the surface representation of the
box is located at the hinge region and the CR7 structure (D). (E and F) Residues comprising
in the P6.1 loop decreased the telomerase ACA box is located at the 3′-end tail (66). the CB localization signal are highlighted in dark
activity in vitro by approximately threefold In addition to the H/ACA motif, scaRNAs blue (CAB box) and light blue in the secondary
but slightly increased the telomere addi- share another common motif, known as structure (E) and colored as in B in the surface
tion processivity (∼20%), indicating that the CAB box, which has a consensus representation of the CR7 structure (F).

20330 | www.pnas.org/cgi/doi/10.1073/pnas.1100279108 Zhang et al.


sequence and structural elements of the revealed that the first two structural fea- tional analysis, structure determination of
two independent signals, located in CR7, tures described above are shared in com- subdomains, and structure-based model-
for 3′-end processing and accumulation mon between these two RNAs, and ing of hTR in conjunction with bio-
and localization of hTR to Cajal bodies additional mutational analysis confirmed chemical characterization have revealed
(Fig. 6) (60). The CR7 hairpin loop has that the processing signal for 3′-end mat- essential elements of hTR structure and
four key structural features. First, within uration of hTR comprises a U-G base pair function beyond the role in templating the
the 8-nt loop, there is 1 bp observed be- between the first nucleotide of the loop telomere DNA. Important insights in-
tween the first and the second to last nu- and the penultimate nucleotide of the clude the identification and character-
cleotides of the loop, a U411·G417 wobble loop, an unpaired U418 at the last position ization of an essential triple helix in the
pair. Second, because of the U·G pair in of the loop, and some sequence specificity conserved pseudoknot in the core do-
the loop and stable Watson–Crick base for the top 2 bp in the helix (Fig. 6 C and main, a structurally conserved bulge
pairs in the helix, the last nucleotide of the D). Comparison of the structures of the loop in the core domain that directs the
loop, U418, adopts an unpaired confor- hTR CR7 hairpin loop and the human overall topology, and two independent
mation. Third, the first 3 loop nt that are U85 C/D-H/ACA scaRNA, combined with signals, for Cajal body localization and
also the first 3 nt of the CAB box are characterization of the processing, locali- hTR 3′-end processing, in the 3′-terminal
stacked over each other, forming A-form zation, and accumulation of various mu- hairpin of hTR. Recent structural studies
backbone geometry. Finally, although the tant and Wt RNAs, revealed that the CB of the core domain have provided a
last CAB box residue (G414) does not localization signal comprises the ugAG
testable model structure of the core do-
stack over other residues and is quite CAB box nucleotides as well as a base-
main that is consistent with all current
flexible, a change in the backbone di- paired helix, with some dependence of the
biochemical data. However, much re-
rection occurs between G414 and C415, sequence identity of the top 2 bp (Fig. 6 E
mains to be learned, particularly about
positioning all CAB box residues on the 5′ and F). Recently, the protein responsible
side of the loop. To determine which of for binding to the CB localization signal how and where hTERT interacts with
these structural features is important for and driving telomerase and other H/ACA the core domain and how the core do-
the 3′-end processing and localization scaRNAs to Cajal bodies has been identi- main and the STE interact with hTERT
signals, parallel structural determinations fied as TCAB1/WDR79 (72, 73). It is in- and potentially each other to facilitate
were performed on the terminal loop teresting to note that, although they are catalysis. The structure of the H/ACA
construct derived from the human U64 H/ both in the same hairpin, the CB locali- scaRNA domain and how two sets of
ACA snoRNA and human U85 C/D-H/ zation signal and the 3′-end processing H/ACA RNP proteins bind two con-
ACA scaRNA. It had been previously signal could potentially independently and secutive hairpin-bulge-hairpin sequences
shown the U64 snoRNA replacement of simultaneously bind TCAB1/WDR79 and remain to be determined. Combined
the hTR CR7 in the full-length hTR al- the unidentified protein that is required use of NMR, X-ray crystallography,
lows hTR accumulation and processing, for TR accumulation and 3′-end process- small-angle X-ray scattering, single mol-
but the RNA localizes to nucleoli instead ing. Furthermore, the 3′ processing signal ecule techniques, and EM will likely re-
of Cajal bodies. In contrast, replacement is the same site where the H/ACA RNP veal these structures within the next
of the CR7 hairpin with the 3′ hairpin of protein Nhp2 would be expected to bind few years.
U85 scaRNA abrogated processing, and (60, 106).
the hTR remained at the site of tran- ACKNOWLEDGMENTS. Q.Z. is a Baltimore Family
Summary Fellow of the Life Sciences Research Foundation.
scription. Comparison of the terminal loop This work was supported by grants from the Na-
structures of the hTR CR7 and the 5′ During the past decade, combined use of tional Institutes of Health and the National Science
hairpin of human U64 H/ACA snoRNA secondary structure predictions, muta- Foundation (to J.F.).

1. Palm W, de Lange T (2008) How shelterin protects 13. Vulliamy T, et al. (2001) The RNA component of telo- 22. Xin ZT, et al. (2007) Functional characterization of
mammalian telomeres. Annu Rev Genet 42:301–334. merase is mutated in autosomal dominant dyskeratosis natural telomerase mutations found in patients with
2. Martínez P, Blasco MA (2011) Telomeric and extra- congenita. Nature 413:432–435. hematologic disorders. Blood 109:524–532.
telomeric roles for telomerase and the telomere- 14. Yamaguchi H, et al. (2003) Mutations of the hu- 23. Marrone A, et al. (2007) Functional characterization
binding proteins. Nat Rev Cancer 11:161–176. man telomerase RNA gene (TERC) in aplastic anemia of novel telomerase RNA (TERC) mutations in patients
3. Wong JM, Collins K (2003) Telomere maintenance and and myelodysplastic syndrome. Blood 102:916– with diverse clinical and pathological presentations.
disease. Lancet 362:983–988. 918. Haematologica 92:1013–1020.
4. Blasco MA (2007) Telomere length, stem cells and 15. Chen JL, Greider CW (2004) An emerging consensus 24. Calado RT, Young NS (2008) Telomere maintenance
aging. Nat Chem Biol 3:640–649. for telomerase RNA structure. Proc Natl Acad Sci USA and human bone marrow failure. Blood 111:
5. Collado M, Blasco MA, Serrano M (2007) Cellu- 101:14683–14684. 4446–4455.
lar senescence in cancer and aging. Cell 130:223–233. 16. Vulliamy T, et al. (2004) Disease anticipation is 25. Armanios M (2009) Syndromes of telomere short-
6. Aubert G, Lansdorp PM (2008) Telomeres and aging. associated with progressive telomere shortening in ening. Annu Rev Genomics Hum Genet 10:45–61.
Physiol Rev 88:557–579. families with dyskeratosis congenita due to mutations 26. Du HY, et al. (2009) TERC and TERT gene mutations in
7. Greider CW, Blackburn EH (1985) Identification of in TERC. Nat Genet 36:447–449. patients with bone marrow failure and the
a specific telomere terminal transferase activity in 17. Marrone A, Walne A, Dokal I (2005) Dyskeratosis significance of telomere length measurements. Blood
Tetrahymena extracts. Cell 43:405–413. congenita: Telomerase, telomeres and anticipation. 113:309–316.
8. Blackburn EH, Greider CW, Szostak JW (2006) Curr Opin Genet Dev 15:249–257. 27. Autexier C, Lue NF (2006) The structure and function
Telomeres and telomerase: The path from maize, 18. Ly H, et al. (2005) Identification and functional of telomerase reverse transcriptase. Annu Rev
Tetrahymena and yeast to human cancer and aging. characterization of 2 variant alleles of the telomerase Biochem 75:493–517.
Nat Med 12:1133–1138. RNA template gene (TERC) in a patient with 28. Collins K (2006) The biogenesis and regulation of
9. Blackburn EH, Collins K (2011) Telomerase: An RNP dyskeratosis congenita. Blood 106:1246–1252. telomerase holoenzymes. Nat Rev Mol Cell Biol 7:
enzyme synthesizes DNA. Cold Spring Harb Perspect 19. Ly H, et al. (2005) Functional characterization of 484–494.
Biol 3:a003558. telomerase RNA variants found in patients with 29. Collins K (2008) Physiological assembly and activity of
10. Blasco MA (2005) Telomeres and human disease: hematologic disorders. Blood 105:2332–2339. human telomerase complexes. Mech Ageing Dev 129:
Ageing, cancer and beyond. Nat Rev Genet 6: 20. Vulliamy TJ, et al. (2006) Mutations in dyskeratosis 91–98.
611–622. congenita: Their impact on telomere length and the 30. Lingner J, et al. (1997) Reverse transcriptase motifs in
11. Shay JW, Wright WE (2006) Telomerase therapeutics diversity of clinical presentation. Blood 107: the catalytic subunit of telomerase. Science 276:
for cancer: Challenges and new directions. Nat Rev 2680–2685. 561–567.
Drug Discov 5:577–584. 21. Armanios MY, et al. (2007) Telomerase mutations in 31. Wyatt HD, West SC, Beattie TL (2010) InTERTpreting
12. Harley CB (2008) Telomerase and cancer therapeutics. families with idiopathic pulmonary fibrosis. N Engl J Med telomerase structure and function. Nucleic Acids Res
Nat Rev Cancer 8:167–179. 356:1317–1326. 38:5609–5622.

Zhang et al. PNAS | December 20, 2011 | vol. 108 | no. 51 | 20331
32. Mason M, Schuller A, Skordalakes E (2011) Telomerase 57. Lukowiak AA, Narayanan A, Li ZH, Terns RM, 82. Yingling YG, Shapiro BA (2006) The prediction of the
structure function. Curr Opin Struct Biol 21:92–100. Terns MP (2001) The snoRNA domain of vertebrate wild-type telomerase RNA pseudoknot structure and
33. Jacobs SA, Podell ER, Cech TR (2006) Crystal structure telomerase RNA functions to localize the RNA within the pivotal role of the bulge in its formation. J Mol
of the essential N-terminal domain of telomerase the nucleus. RNA 7:1833–1844. Graph Model 25:261–274.
reverse transcriptase. Nat Struct Mol Biol 13:218–225. 58. Fu D, Collins K (2003) Distinct biogenesis pathways for 83. Chen JL, Greider CW (2003) Determinants in
34. Zaug AJ, Podell ER, Cech TR (2008) Mutation in TERT human telomerase RNA and H/ACA small nucleolar mammalian telomerase RNA that mediate enzyme
separates processivity from anchor-site function. Nat RNAs. Mol Cell 11:1361–1372. processivity and cross-species incompatibility. EMBO J
Struct Mol Biol 15:870–872. 59. Jády BE, Bertrand E, Kiss T (2004) Human telomerase 22:304–314.
35. Rouda S, Skordalakes E (2007) Structure of the RNA- RNA and box H/ACA scaRNAs share a common Cajal 84. Popenda M, et al. (2010) RNA FRABASE 2.0: An
binding domain of telomerase: Implications for RNA body-specific localization signal. J Cell Biol 164: advanced web-accessible database with the capacity
recognition and binding. Structure 15:1403–1412. 647–652. to search the three-dimensional fragments within
36. Gillis AJ, Schuller AP, Skordalakes E (2008) Structure 60. Theimer CA, et al. (2007) Structural and functional RNA structures. BMC Bioinformatics 11:231.
of the Tribolium castaneum telomerase catalytic characterization of human telomerase RNA 85. Lukavsky PJ, Kim I, Otto GA, Puglisi JD (2003)
subunit TERT. Nature 455:633–637. processing and cajal body localization signals. Mol Structure of HCV IRES domain II determined by NMR.
37. Mitchell M, Gillis A, Futahashi M, Fujiwara H, Cell 27:869–881. Nat Struct Biol 10:1033–1038.
Skordalakes E (2010) Structural basis for telomerase 61. Cristofari G, et al. (2007) Human telomerase RNA 86. Luebke KJ, Landry SM, Tinoco I, Jr. (1997) Solution
catalytic subunit TERT binding to RNA template and accumulation in Cajal bodies facilitates telomerase conformation of a five-nucleotide RNA bulge loop
telomeric DNA. Nat Struct Mol Biol 17:513–518. recruitment to telomeres and telomere elongation. from a group I intron. Biochemistry 36:10246–10255.
38. Greider CW, Blackburn EH (1989) A telomeric Mol Cell 27:882–889. 87. Jenner LB, Demeshkina N, Yusupova G, Yusupov M
sequence in the RNA of Tetrahymena telomerase 62. Chen JL, Greider CW (2004) Telomerase RNA structure (2010) Structural aspects of messenger RNA reading
required for telomere repeat synthesis. Nature 337: and function: Implications for dyskeratosis congenita. frame maintenance by the ribosome. Nat Struct Mol
331–337. Trends Biochem Sci 29:183–192. Biol 17:555–560.
39. Podlevsky JD, Bley CJ, Omana RV, Qi X, Chen JJ (2008) 63. Ly H, Blackburn EH, Parslow TG (2003) Comprehensive 88. Palmer AG, 3rd (2004) NMR characterization of the
The telomerase database. Nucleic Acids Res 36: structure-function analysis of the core domain of dynamics of biomacromolecules. Chem Rev 104:
D339–D343. human telomerase RNA. Mol Cell Biol 23:6849–6856. 3623–3640.
40. Gunisova S, et al. (2009) Identification and comparative 64. Chen JL, Opperman KK, Greider CW (2002) A critical 89. Tjandra N, Bax A (1997) Direct measurement of
analysis of telomerase RNAs from Candida species stem-loop structure in the CR4-CR5 domain of distances and angles in biomolecules by NMR in a dilute
reveal conservation of functional elements. RNA 15: mammalian telomerase RNA. Nucleic Acids Res 30: liquid crystalline medium. Science 278:1111–1114.
546–559. 592–597. 90. Prestegard JH, al-Hashimi HM, Tolman JR (2000) NMR
41. Kachouri-Lafond R, et al. (2009) Large telomerase 65. Zhu Y, Tomlinson RL, Lukowiak AA, Terns RM, structures of biomolecules using field oriented media
RNA, telomere length heterogeneity and escape from Terns MP (2004) Telomerase RNA accumulates in Cajal and residual dipolar couplings. Q Rev Biophys 33:
senescence in Candida glabrata. FEBS Lett 583: bodies in human cancer cells. Mol Biol Cell 15:81–90. 371–424.
3605–3610. 66. Kiss T, Fayet-Lebaron E, Jády BE (2010) Box H/ACA 91. Zhang Q, Stelzer AC, Fisher CK, Al-Hashimi HM (2007)
42. Romero DP, Blackburn EH (1991) A conserved small ribonucleoproteins. Mol Cell 37:597–606. Visualizing spatially correlated dynamics that directs
secondary structure for telomerase RNA. Cell 67: 67. Egan ED, Collins K (2010) Specificity and stoichiometry RNA conformational transitions. Nature 450:1263–1267.
343–353. of subunit interactions in the human telomerase 92. Parisien M, Major F (2008) The MC-Fold and MC-Sym
43. McCormick-Graham M, Romero DP (1995) Ciliate holoenzyme assembled in vivo. Mol Cell Biol 30: pipeline infers RNA structure from sequence data.
telomerase RNA structural features. Nucleic Acids Res 2775–2786. Nature 452:51–55.
23:1091–1097. 68. Li L, Ye K (2006) Crystal structure of an H/ACA box 93. Gavory G, Symmons MF, Krishnan Ghosh Y,
44. McCormick-Graham M, Romero DP (1996) A single ribonucleoprotein particle. Nature 443:302–307. Klenerman D, Balasubramanian S (2006) Structural
telomerase RNA is sufficient for the synthesis of 69. Liang B, Xue S, Terns RM, Terns MP, Li H (2007) Substrate analysis of the catalytic core of human telomerase
variable telomeric DNA repeats in ciliates of the RNA positioning in the archaeal H/ACA ribonucleo- RNA by FRET and molecular modeling. Biochemistry
genus Paramecium. Mol Cell Biol 16:1871–1879. protein complex. Nat Struct Mol Biol 14:1189–1195. 45:13304–13311.
45. Ye AJ, Romero DP (2002) Phylogenetic relationships 70. Liang B, et al. (2009) Structure of a functional 94. Qiao F, Goodrich KJ, Cech TR (2010) Engineering cis-
amongst tetrahymenine ciliates inferred by a com- ribonucleoprotein pseudouridine synthase bound to telomerase RNAs that add telomeric repeats to
parison of telomerase RNAs. Int J Syst Evol Microbiol a substrate RNA. Nat Struct Mol Biol 16:740–746. themselves. Proc Natl Acad Sci USA 107:4914–4918.
52:2297–2302. 71. Duan J, Li L, Lu J, Wang W, Ye K (2009) Structural 95. Lai CK, Miller MC, Collins K (2003) Roles for RNA in
46. Chen JL, Blasco MA, Greider CW (2000) Secondary mechanism of substrate RNA recruitment in H/ACA telomerase nucleotide and repeat addition
structure of vertebrate telomerase RNA. Cell 100: RNA-guided pseudouridine synthase. Mol Cell 34: processivity. Mol Cell 11:1673–1683.
503–514. 427–439. 96. Mason DX, Goneska E, Greider CW (2003) Stem-loop
47. Xie M, et al. (2008) Structure and function of the 72. Tycowski KT, Shu MD, Kukoyi A, Steitz JA (2009) A IV of tetrahymena telomerase RNA stimulates pro-
smallest vertebrate telomerase RNA from teleost fish. conserved WD40 protein binds the Cajal body cessivity in trans. Mol Cell Biol 23:5606–5613.
J Biol Chem 283:2049–2059. localization signal of scaRNP particles. Mol Cell 34:47–57. 97. Brown Y, et al. (2007) A critical three-way junction is
48. Theimer CA, Feigon J (2006) Structure and function of 73. Venteicher AS, et al. (2009) A human telomerase conserved in budding yeast and vertebrate
telomerase RNA. Curr Opin Struct Biol 16:307–318. holoenzyme protein required for Cajal body localization telomerase RNAs. Nucleic Acids Res 35:6280–6289.
49. Autexier C, Pruzan R, Funk WD, Greider CW (1996) and telomere synthesis. Science 323:644–648. 98. Robart AR, Collins K (2010) Investigation of human
Reconstitution of human telomerase activity and 74. Leeper TC, Varani G (2005) The structure of an telomerase holoenzyme assembly, activity, and
identification of a minimal functional region of the enzyme-activating fragment of human telomerase processivity using disease-linked subunit variants.
human telomerase RNA. EMBO J 15:5928–5935. RNA. RNA 11:394–403. J Biol Chem 285:4375–4386.
50. Mitchell JR, Collins K (2000) Human telomerase 75. Leeper T, Leulliot N, Varani G (2003) The solution 99. Ueda CT, Roberts RW (2004) Analysis of a long-range
activation requires two independent interactions structure of an essential stem-loop of human interaction between conserved domains of human
between telomerase RNA and telomerase reverse telomerase RNA. Nucleic Acids Res 31:2614–2621. telomerase RNA. RNA 10:139–147.
transcriptase. Mol Cell 6:361–371. 76. Kim NK, Theimer CA, Mitchell JR, Collins K, Feigon J 100. Mueller EG, Ferre-D’Amare AR (2009) Pseudouridine
51. Martin-Rivera L, Blasco MA (2001) Identification of (2010) Effect of pseudouridylation on the structure formation, the most common transglycosylation in
functional domains and dominant negative mutations and activity of the catalytically essential P6.1 hairpin RNA. DNA and RNA Modification Enzymes: Structure,
in vertebrate telomerase RNA using an in vivo in human telomerase RNA. Nucleic Acids Res 38: Mechanism, Function and Evolution, ed Grosjean H
reconstitution system. J Biol Chem 276:5856–5865. 6746–6756. (Landes Bioscience, Austin, TX), pp 363–376.
52. Theimer CA, Blois CA, Feigon J (2005) Structure of the 77. Theimer CA, Finger LD, Trantirek L, Feigon J (2003) 101. Ganot P, Bortolin ML, Kiss T (1997) Site-specific
human telomerase RNA pseudoknot reveals Mutations linked to dyskeratosis congenita cause pseudouridine formation in preribosomal RNA is
conserved tertiary interactions essential for function. changes in the structural equilibrium in telomerase guided by small nucleolar RNAs. Cell 89:799–809.
Mol Cell 17:671–682. RNA. Proc Natl Acad Sci USA 100:449–454. 102. Ni J, Tien AL, Fournier MJ (1997) Small nucleolar RNAs
53. Qiao F, Cech TR (2008) Triple-helix structure in 78. Comolli LR, Smirnov I, Xu L, Blackburn EH, James TL direct site-specific synthesis of pseudouridine in
telomerase RNA contributes to catalysis. Nat Struct (2002) A molecular switch underlies a human ribosomal RNA. Cell 89:565–573.
Mol Biol 15:634–640. telomerase disease. Proc Natl Acad Sci USA 99: 103. Smith CM, Steitz JA (1997) Sno storm in the nucleolus:
54. Kim NK, et al. (2008) Solution structure and dynamics 16998–17003. New roles for myriad small RNPs. Cell 89:669–672.
of the wild-type pseudoknot of human telomerase 79. Chen JL, Greider CW (2005) Functional analysis of the 104. Darzacq X, et al. (2002) Cajal body-specific small
RNA. J Mol Biol 384:1249–1261. pseudoknot structure in human telomerase RNA. Proc nuclear RNAs: A novel class of 2′-O-methylation and
55. Zhang Q, Kim NK, Peterson RD, Wang Z, Feigon J Natl Acad Sci USA 102:8080–8085. pseudouridylation guide RNAs. EMBO J 21:2746–2756.
(2010) Structurally conserved five nucleotide bulge 80. Shefer K, et al. (2007) A triple helix within 105. Richard P, et al. (2003) A common sequence motif
determines the overall topology of the core domain a pseudoknot is a conserved and essential element of determines the Cajal body-specific localization of box
of human telomerase RNA. Proc Natl Acad Sci USA telomerase RNA. Mol Cell Biol 27:2130–2143. H/ACA scaRNAs. EMBO J 22:4283–4293.
107:18761–18768. 81. Ulyanov NB, Shefer K, James TL, Tzfati Y (2007) 106. Koo BK, et al. (2011) Structure of H/ACA RNP protein
56. Mitchell JR, Cheng J, Collins K (1999) A box H/ACA Pseudoknot structures with conserved base triples in Nhp2p reveals cis/trans isomerization of a conserved
small nucleolar RNA-like domain at the human telomerase RNAs of ciliates. Nucleic Acids Res 35: proline at the RNA and Nop10 binding interface.
telomerase RNA 3′ end. Mol Cell Biol 19:567–576. 6150–6160. J Mol Biol, 10.1016/j.jmb.2011.06.022.

20332 | www.pnas.org/cgi/doi/10.1073/pnas.1100279108 Zhang et al.

You might also like